Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Cell ; 187(11): 2817-2837.e31, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38701783

ABSTRACT

FMS-related tyrosine kinase 3 ligand (FLT3L), encoded by FLT3LG, is a hematopoietic factor essential for the development of natural killer (NK) cells, B cells, and dendritic cells (DCs) in mice. We describe three humans homozygous for a loss-of-function FLT3LG variant with a history of various recurrent infections, including severe cutaneous warts. The patients' bone marrow (BM) was hypoplastic, with low levels of hematopoietic progenitors, particularly myeloid and B cell precursors. Counts of B cells, monocytes, and DCs were low in the patients' blood, whereas the other blood subsets, including NK cells, were affected only moderately, if at all. The patients had normal counts of Langerhans cells (LCs) and dermal macrophages in the skin but lacked dermal DCs. Thus, FLT3L is required for B cell and DC development in mice and humans. However, unlike its murine counterpart, human FLT3L is required for the development of monocytes but not NK cells.


Subject(s)
Killer Cells, Natural , Membrane Proteins , Animals , Female , Humans , Male , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/cytology , Bone Marrow/metabolism , Cell Lineage , Dendritic Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Langerhans Cells/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Monocytes/metabolism , Skin/metabolism , Mice, Inbred C57BL
2.
Cell ; 186(23): 5114-5134.e27, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37875108

ABSTRACT

Human inherited disorders of interferon-gamma (IFN-γ) immunity underlie severe mycobacterial diseases. We report X-linked recessive MCTS1 deficiency in men with mycobacterial disease from kindreds of different ancestries (from China, Finland, Iran, and Saudi Arabia). Complete deficiency of this translation re-initiation factor impairs the translation of a subset of proteins, including the kinase JAK2 in all cell types tested, including T lymphocytes and phagocytes. JAK2 expression is sufficiently low to impair cellular responses to interleukin-23 (IL-23) and partially IL-12, but not other JAK2-dependent cytokines. Defective responses to IL-23 preferentially impair the production of IFN-γ by innate-like adaptive mucosal-associated invariant T cells (MAIT) and γδ T lymphocytes upon mycobacterial challenge. Surprisingly, the lack of MCTS1-dependent translation re-initiation and ribosome recycling seems to be otherwise physiologically redundant in these patients. These findings suggest that X-linked recessive human MCTS1 deficiency underlies isolated mycobacterial disease by impairing JAK2 translation in innate-like adaptive T lymphocytes, thereby impairing the IL-23-dependent induction of IFN-γ.


Subject(s)
Interferon-gamma , Janus Kinase 2 , Mycobacterium Infections , Humans , Male , Cell Cycle Proteins/metabolism , Interferon-gamma/immunology , Interleukin-12 , Interleukin-23 , Janus Kinase 2/metabolism , Mycobacterium/physiology , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Oncogene Proteins/metabolism
3.
Nat Immunol ; 25(5): 764-777, 2024 May.
Article in English | MEDLINE | ID: mdl-38609546

ABSTRACT

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.


Subject(s)
Immunologic Deficiency Syndromes , Nerve Tissue Proteins , Ubiquitins , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Female , Male , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/genetics , Inflammation/immunology , Inflammation/genetics , B-Lymphocytes/immunology , Loss of Function Mutation , Fibroblasts/metabolism , Fibroblasts/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Mice , Alleles
4.
Nature ; 623(7988): 803-813, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938781

ABSTRACT

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Subject(s)
Autoantibodies , Genetic Predisposition to Disease , Interferon Type I , NF-kappa B , Humans , Autoantibodies/immunology , COVID-19/genetics , COVID-19/immunology , Gain of Function Mutation , Heterozygote , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Loss of Function Mutation , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p52 Subunit/deficiency , NF-kappa B p52 Subunit/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology , Thymus Gland/pathology , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , AIRE Protein , NF-kappaB-Inducing Kinase
5.
J Clin Immunol ; 43(8): 1941-1952, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37584719

ABSTRACT

PURPOSE: Major histocompatibility complex class II (MHC-II) deficiency is a rare inborn error of immunity (IEI). Impaired antigen presentation to CD4 + T cells results in combined immunodeficiency (CID). Patients typically present with severe respiratory and gastrointestinal tract infections at early ages. Hematopoietic stem cell transplantation (HSCT) is the only curative therapy. METHODS: We describe the clinical, immunologic, and genetic features of eighteen unrelated Iranian patients with MHC-II deficiency. RESULTS: Consanguinity was present in all affected families. The median age at the initial presentation was 5.5 months (range 7 days to 18 years). The main symptoms included failure to thrive, persistent diarrhea, and pneumonia. Autoimmune and neurologic features were also documented in about one-third of the patients, respectively. Thirteen patients carried RFXANK gene mutations, two carried RFX5 gene mutations, and three carried a RFXAP gene mutation. Six patients shared the same RFXANK founder mutation (c.162delG); limited to the Iranian population and dated to approximately 1296 years ago. Four of the patients underwent HSCT; three of them are alive. On the other hand, nine of the fourteen patients who did not undergo HSCT had a poor prognosis and died. CONCLUSION: MHC-II deficiency is not rare in Iran, with a high rate of consanguinity. It should be considered in the differential diagnosis of CID at any age. With the limited access to HSCT and its variable results in MHC-II deficiency, implementing genetic counseling and family planning for the affected families are mandatory. We are better determined to study the c.162delG RFXANK heterozygous mutation frequency in the Iranian population.


Subject(s)
DNA-Binding Proteins , Severe Combined Immunodeficiency , Transcription Factors , Humans , Infant, Newborn , DNA-Binding Proteins/genetics , Histocompatibility Antigens Class II/genetics , Iran , Mutation/genetics , Severe Combined Immunodeficiency/genetics , Transcription Factors/genetics
6.
Blood Cells Mol Dis ; 99: 102726, 2023 03.
Article in English | MEDLINE | ID: mdl-36696755

ABSTRACT

Leukocyte adhesion deficiency (LAD) is an immunodeficiency caused by defects in the adhesion of leukocytes (especially neutrophils) to the blood vessel wall. As a result, patients with LAD suffer from severe bacterial infections and impaired wound healing, accompanied by neutrophilia. In LAD-I, characterized directly after birth by delayed separation of the umbilical cord, mutations are found in ITGB2, the gene that encodes the ß subunit (CD18) of the ß2 integrins. In the rare LAD-II disease, the fucosylation of selectin ligands is disturbed, caused by mutations in SLC35C1, the gene that encodes a GDP-fucose transporter of the Golgi system. LAD-II patients lack the H and Lewis Lea and Leb blood group antigens. Finally, in LAD-III, the conformational activation of the hematopoietically expressed ß integrins is disturbed, leading to leukocyte and platelet dysfunction. This last syndrome is caused by mutations in FERMT3, encoding the kindlin-3 protein in all blood cells, involved in the regulation of ß integrin conformation. This article contains an update of the mutations that we consider to be relevant for the various forms of LAD.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , Humans , Cell Adhesion/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , CD18 Antigens/genetics , CD18 Antigens/metabolism , Leukocytes , Mutation
7.
Pediatr Allergy Immunol ; 34(7): e13990, 2023 07.
Article in English | MEDLINE | ID: mdl-37492921

ABSTRACT

BACKGROUND: In order to support the comprehensive classification of Leukocyte Adhesion Deficiency-I (LAD-I) severity by simultaneous screening of CD11a/CD18, this study assessed clinical, laboratory, and genetic findings along with outcomes of 69 LAD-I patients during the last 15 years. METHODS: Sixty-nine patients (40 females and 29 males) with a clinical phenotype suspected of LAD-I were referred to Immunology, Asthma, and Allergy research institute, Tehran, Iran between 2007 and 2022 for further advanced immunological screening and genetic evaluations as well as treatment, were enrolled in this study. RESULTS: The diagnosis median age of the patients was 6 months. Delayed umbilical cord separation was found in 25 patients (36.2%). The median diagnostic delay time was 4 months (min-max: 0-82 months). Forty-six patients (66.7%) were categorized as severe (CD18 and/or CD11a: below 2%); while 23 children (33.3%) were in moderate category (CD18 and/or CD11a: 2%-30%). During the follow-ups, 55.1% of children were alive with a mortality rate of 44.9%. Skin ulcers (75.4%), omphalitis (65.2%), and gingivitis (37.7%) were the most frequent complaints. Genetic analysis of the patients revealed 14 previously reported and three novel pathogenic mutations in the ITGB2 gene. The overall survival of patients with and without hematopoietic stem cell transplantation was 79.3% and 55.6%, respectively. CONCLUSION: Physicians' awareness of LAD-I considering delayed separation of umbilical cord marked neutrophilic leukocytosis, and variability in CD11 and CD18 expression levels, and genetic analysis leads to early diagnosis and defining disease severity. Moreover, the prenatal diagnosis would benefit families with a history of LAD-I.


Subject(s)
CD18 Antigens , Leukocyte-Adhesion Deficiency Syndrome , Male , Pregnancy , Female , Humans , CD18 Antigens/genetics , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , Leukocyte-Adhesion Deficiency Syndrome/genetics , Delayed Diagnosis , Iran , Leukocytes/metabolism
8.
Fetal Pediatr Pathol ; 42(2): 259-262, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35608383

ABSTRACT

INTRODUCTION: Chediak-Higashi syndrome (CHS) is caused by dysfunction of lysosomal trafficking and presents with hypopigmentation, bleeding tendencies, neurological symptoms, and NK cell dysfunction. Hemophagocytic lymphohistiocytosis (HLH) can complicate CHS due to the abnormal function of NK cells. CASE PRESENTATION: This 1.5-year-old light-skinned gray-haired girl microscopically had abnormal hair pigment clumps and lilac inclusions in the myeloid series, characteristic of CHS. She presented with HLH, requiring treatment with etoposide and dexamethasone followed by cyclosporine and dexamethasone. CONCLUSION: CHS is one of the underlying primary causes of HLH.


Subject(s)
Chediak-Higashi Syndrome , Lymphohistiocytosis, Hemophagocytic , Female , Humans , Infant , Chediak-Higashi Syndrome/complications , Chediak-Higashi Syndrome/diagnosis , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/diagnosis , Skin , Vesicular Transport Proteins , Dexamethasone/therapeutic use
9.
Ann Clin Microbiol Antimicrob ; 21(1): 8, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35232430

ABSTRACT

BACKGROUND: Post-vaccination BCG disease typically attests to underlying inborn errors of immunity (IEIs), with the highest rates of complications in patients with Mendelian susceptibility to mycobacterial disease (MSMD). However, therapeutic protocols for the management of BCG-osis (disseminated) and persistent BCG-itis (localized) are still controversial. METHODS: Twenty-four Iranian patients with MSMD (BCG-osis or BCG-itis), followed from 2009 to 2020 in Tehran, were included in the study. Their medical records were retrospectively reviewed for demographics, clinical features, laboratory findings, and molecular diagnosis. The therapeutic protocol sheets were prepared to contain the types and duration of anti-mycobacterial agents. RESULTS: BCG disease either as BCG-itis (33.3%) or BCG-osis (66.7%) was confirmed in all patients by positive gastric washing test (54.2%), microbial smear and culture (58.3%), or purified protein derivative (PPD) test (4.2%). The duration between BCG-osis onset and MSMD diagnosis was 21.6 months. All except three patients were initiated on second-line anti-mycobacterial agents with either a fluoroquinolone (levofloxacin: 15 mg/kg/day, ciprofloxacin: 20 mg/kg/day, ofloxacin: 15 mg/kg/day), aminoglycoside (amikacin: 10-15 mg/kg/day, streptomycin: 15 mg/kg/day), and/or macrolide (clarithromycin: 15 mg/kg/day) along with oral rifampin (10 mg/kg/day), isoniazid (15 mg/kg/day), and ethambutol (20 mg/kg/day). Three patients showed a clinical response to rifampin, despite in vitro resistance. Fourteen (58.3%) patients received also adjuvant subcutaneous IFN-γ therapy, 50 µ/m2 every other day. At the end of survey, most patients (n = 22, 91.7%) were alive and two patients died following BCG-osis and respiratory failure. CONCLUSIONS: We recommend the early instigation of second-line anti-mycobacterial agents in MSMD patients with BCG disease.


Subject(s)
BCG Vaccine , Mycobacterium Infections , BCG Vaccine/therapeutic use , Genetic Predisposition to Disease , Humans , Iran , Mycobacterium Infections/diagnosis , Mycobacterium Infections/drug therapy , Retrospective Studies
10.
J Wound Care ; 31(5): 427-431, 2022 May 02.
Article in English | MEDLINE | ID: mdl-35579316

ABSTRACT

Patients with chronic granulomatous disease, a primary immunodeficiency, experience granulomatous complications and recurrent life-threatening opportunistic bacterial and fungal infections. In this article, we report on a case of invasive aspergillosis in an eight-year-old boy with chronic granulomatous disease, who presented with pleural effusion and pneumonia, cerebral venous sinus thrombosis, and unusual skin lesions caused by Aspergillus fumigatus. Antifungal treatment with itraconazole and other antifungal agents, along with interferon-γ, was ineffective and the patient eventually died from cerebral venous sinus thrombosis, and intracerebral haemorrhage following increased intracranial pressure after one month. The diagnosis of invasive aspergillosis should be considered early in children presenting with invasive fungal infections, particularly those involving the central nervous system.


Subject(s)
Aspergillosis , Granulomatous Disease, Chronic , Invasive Fungal Infections , Sinus Thrombosis, Intracranial , Antifungal Agents/therapeutic use , Aspergillosis/complications , Aspergillosis/diagnosis , Aspergillosis/drug therapy , Child , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/diagnosis , Granulomatous Disease, Chronic/microbiology , Humans , Invasive Fungal Infections/complications , Invasive Fungal Infections/drug therapy , Itraconazole/therapeutic use , Male , Sinus Thrombosis, Intracranial/complications , Sinus Thrombosis, Intracranial/drug therapy
11.
Rheumatology (Oxford) ; 60(11): 5436-5446, 2021 11 03.
Article in English | MEDLINE | ID: mdl-33693560

ABSTRACT

OBJECTIVE: In 2016 specific heterozygous gain-of-function mutations in the Mediterranean fever gene MEFV were reported as causal for a distinct autoinflammatory disease coined pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). We sought to provide an extended report on clinical manifestations in PAAND patients to date and evaluate the efficacy and safety of treatment with the IL-1-blocking agent anakinra. METHODS: We undertook an open-label pilot study with anakinra. Three patients were recruited in a preliminary phase of the study with the intention to expand the treatment cohort in case of a favourable response. Acute-phase reactants and plasma cytokine levels were monitored throughout. Skin biopsies at baseline and at week 12 were stained for relevant cytokines. Available clinical data on treatment responses were retrospectively collected on additional patients. RESULTS: The three patients from the preliminary phase of the study [patients 1-3 (P1-P3)] demonstrated one failed and two partial treatment responses, where one patient opted to continue treatment with anakinra and the other favoured adalimumab. While a partial systemic response was observed, there was no appreciable effect of anakinra on the prominent cutaneous manifestations, reflected in residual local inflammatory cytokine expression in lesional skin. These observations did not warrant further expansion of the treatment cohort. Clinical data was retrospectively collected on an additional eight patients (P4-P11), highlighting both dominant and recessive inheritance with variable penetrance in PAAND and common gastrointestinal involvement that was not previously appreciated. CONCLUSION: In our experience, while anakinra appears safe, it was not superior to biologicals targeting TNF-α in PAAND despite evidence directly implicating dysregulated IL-1ß signalling.


Subject(s)
Antirheumatic Agents/therapeutic use , Hereditary Autoinflammatory Diseases/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Leukocyte Disorders/congenital , Skin Diseases, Genetic/drug therapy , Aged , Case-Control Studies , Female , Humans , Leukocyte Disorders/drug therapy , Male , Middle Aged , Phenotype , Pilot Projects , Pyrin/genetics
12.
Allergol Immunopathol (Madr) ; 48(5): 424-429, 2020.
Article in English | MEDLINE | ID: mdl-32653225

ABSTRACT

INTRODUCTION AND OBJECTIVES: Considering that no studies have been done on a comprehensive review of Serum sickness-like reactions patients (SSLRs) at a referral center in Iran so far, this study aimed to determine the clinical and laboratory characteristics of children with SSRL in Tehran Children's Medical Center. PATIENTS: The present study was a registry-based study in which the data of 94 SSLRs patients registered in a two-year period were investigated. Confirmation of fever, rash, urticaria, arthralgia / arthritis and history of antibiotic consumption up to three weeks before were the criteria for the diagnosis. RESULTS: Fifty-one (54 %) patients were male with mean age of 56 ±â€¯30 months and there was no significant difference in the age of the two genders. The mean onset of symptoms before hospitalization were 3.8 ±â€¯2.7 days (1-14 days); this mean was significantly higher in males than in females (4.6 ±â€¯2.9 versus 2.9 ±â€¯1.7 days, P-value = 0.003). Among antibiotics, Co-amoxiclav and Cefixime antibiotics had the most frequency by 31 % and 33 %, respectively as the most important incidence factor of SSLRs. The mean duration of consumption of culprit medications in the incidence of SSLRs was 5.6 ±â€¯2.9 days with a range of 1-15 days. CONCLUSIONS: This study showed that among the antibiotics, Co-amoxiclav and Cefixime are more prevalent and a review of prescribing these two antibiotics for the treatment of the children's infections is essential if this finding is confirmed by other Iranian scholars.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bacterial Infections/drug therapy , Serum Sickness/epidemiology , Amoxicillin-Potassium Clavulanate Combination/adverse effects , Cefixime/adverse effects , Child , Child, Preschool , Female , Humans , Incidence , Iran/epidemiology , Male , Prevalence , Registries/statistics & numerical data , Serum Sickness/etiology , Sex Factors
13.
Emerg Infect Dis ; 25(11): 2005-2012, 2019 11.
Article in English | MEDLINE | ID: mdl-31625840

ABSTRACT

Patients with immunodeficiency-associated vaccine-derived poliovirus (iVDPV) are potential poliovirus reservoirs in the posteradication era that might reintroduce polioviruses into the community. We update the iVDPV registry in Iran by reporting 9 new patients. In addition to national acute flaccid paralysis surveillance, cases were identified by screening nonparalyzed primary immunodeficiency (PID) patients. Overall, 23 iVDPV patients have been identified since 1995. Seven patients (30%) never had paralysis. Poliovirus screening accelerated the iVDPV detection rate in Iran after 2014.The iVDPV infection rate among nonparalyzed patients with adaptive PID was 3.1% (7/224), several folds higher than previous estimates. Severe combined immunodeficiency patients had the highest risk for asymptomatic infection (28.6%) compared with other PIDs. iVDPV2 emergence has decreased after the switch from trivalent to bivalent oral poliovirus vaccine in 2016. However, emergence of iVDPV1 and iVDPV3 continued. Poliovirus screening in PID patients is an essential step in the endgame of polio eradication.


Subject(s)
Poliomyelitis/epidemiology , Poliomyelitis/etiology , Poliovirus Vaccines/adverse effects , Poliovirus/immunology , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/epidemiology , Adolescent , Adult , Asymptomatic Diseases , Child , Child, Preschool , Female , Humans , Infant , Iran/epidemiology , Male , Mass Screening , Outcome Assessment, Health Care , Poliomyelitis/prevention & control , Poliomyelitis/virology , Poliovirus Vaccines/immunology , Primary Immunodeficiency Diseases/therapy , Public Health Surveillance , Registries , Symptom Assessment , Vaccination , Young Adult
14.
Int Arch Allergy Immunol ; 180(1): 52-63, 2019.
Article in English | MEDLINE | ID: mdl-31117086

ABSTRACT

BACKGROUND: Hyper-immunoglobulin M (HIGM) syndrome is a rare heterogeneous group of primary immunodeficiency disorders characterized by low or absent serum levels of IgG and IgA along with normal or elevated serum levels of IgM. METHODS: Clinical and immunological data were collected from the 75 patients' medical records diagnosed in Children's Medical Center affiliated to Tehran University Medical Sciences and other Universities of Medical Sciences in Iran. Among 75 selected patients, 48 patients (64%) were analyzed genetically using targeted and whole-exome sequencing. RESULTS: The ratio of male to female was 2.9:1. The median age at the onset of the disease, time of diagnosis, and diagnostic delay were 10.5, 50, and 24 months, respectively. Pneumonia and lower respiratory tract infections (61.3%) were the most common complications. Responsible genes were identified in 35 patients (72.9%) out 48 genetically analyzed patients. Cluster of differentiation 40 ligand gene was the most mutated gene observed in 24 patients (68.5%) followed by activation-induced cytidine deaminase gene in 7 patients, lipopolysaccharide-responsive and beige-like anchor (1 patient), nuclear factor-kappa-B essential modulator (1 patient), phosphoinositide-3-kinase regulatory subunit 1 (1 patient), and nuclear factor kappa B subunit 1 (1 patient) genes. Nineteen (25.3%) patients died during the study period, and pneumonia was the major cause of death occurred in 6 (31.6%) patients. CONCLUSION: Physicians in our country should carefully pay attention to respiratory tract infections and pneumonia, particularly in patients with a positive family history. Further investigations are required for detection of new genes and pathways resulting in HIGM phenotype.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome/diagnosis , Hyper-IgM Immunodeficiency Syndrome/etiology , Phenotype , Adolescent , Adult , Biomarkers , Child , Disease Susceptibility , Female , Genetic Testing , Humans , Immunoglobulin Isotypes/blood , Iran , Lymphocyte Count , Male , Mutation , Symptom Assessment , Young Adult
15.
Acta Microbiol Immunol Hung ; 67(1): 56-60, 2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31847541

ABSTRACT

Chronic granulomatous disease is a non-prevalent genetic disorder due to different structural gene mutations encoding components of nicotinamide adenine dinucleotide phosphate oxidase complex. Nicotinamide adenine dinucleotide phosphate oxidase is a complex made by a group of five proteins (subunit) and plays an important role in the innate immune system. Five structural genes are responsible for encoding each subunit, in which cytochrome b-245 alpha chain (also known as p22-phox) is encoded by CYBA gene. CYBA gene mutation leads to a group of autosomal dominant chronic granulomatous disease. Decreased level or lack of nicotinamide adenine dinucleotide phosphate oxidase leaves affected individuals vulnerable to many types of infections and excessive inflammation. In this study, a family affected by BCGitis caused by a novel intronic autosomal recessive CYBA mutation (88,713,158 C > T) has been described. The proband is a 5-year-old girl with chronic granulomatous disease who was referred to the clinic due to BCGitis. The culprit mutation was detected following whole genome sequencing and was confirmed among the family members by Sanger sequencing. Being symptom-free at the time of diagnosis, despite the proband's mother homozygosity, was a characteristic feature of this report. Remarkably, none of the CYBA-mutated members, as a known chronic granulomatous disease causing gene, has expressed symptoms other than regional lymph node enlargements. This might explain the gene mutation site importance in demonstrating different manifestations.


Subject(s)
Family , Genes, Recessive , Granulomatous Disease, Chronic/diagnosis , Granulomatous Disease, Chronic/genetics , NADPH Oxidases/genetics , Adult , Child , Child, Preschool , Female , Genetic Testing , Humans , Lymph Nodes/pathology , Male , Mutation , Whole Genome Sequencing
16.
Immunol Rev ; 264(1): 103-20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703555

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb) and a few related mycobacteria, is a devastating disease, killing more than a million individuals per year worldwide. However, its pathogenesis remains largely elusive, as only a small proportion of infected individuals develop clinical disease either during primary infection or during reactivation from latency or secondary infection. Subacute, hematogenous, and extrapulmonary disease tends to be more frequent in infants, children, and teenagers than in adults. Life-threatening primary TB of childhood can result from known acquired or inherited immunodeficiencies, although the vast majority of cases remain unexplained. We review here the conditions conferring a predisposition to childhood clinical diseases caused by mycobacteria, including not only M.tb but also weakly virulent mycobacteria, such as BCG vaccines and environmental mycobacteria. Infections with weakly virulent mycobacteria are much rarer than TB, but the inherited and acquired immunodeficiencies underlying these infections are much better known. Their study has also provided genetic and immunological insights into childhood TB, as illustrated by the discovery of single-gene inborn errors of IFN-γ immunity underlying severe cases of TB. Novel findings are expected from ongoing and future human genetic studies of childhood TB in countries that combine a high proportion of consanguineous marriages, a high incidence of TB, and an excellent clinical care, such as Iran, Morocco, and Turkey.


Subject(s)
Disease Susceptibility/immunology , Genetic Predisposition to Disease , Immunocompromised Host , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/etiology , Mycobacterium tuberculosis/immunology , Tuberculosis/etiology , Age Factors , Child , Genes, Dominant , Genes, Recessive , Humans , Immunologic Deficiency Syndromes/diagnosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
J Clin Immunol ; 38(5): 617-627, 2018 07.
Article in English | MEDLINE | ID: mdl-29995221

ABSTRACT

PURPOSE: Inborn errors of IFN-γ immunity underlie Mendelian susceptibility to mycobacterial disease (MSMD). Autosomal recessive complete IL-12Rß1 deficiency is the most frequent genetic etiology of MSMD. Only two of the 84 known mutations are copy number variations (CNVs), identified in two of the 213 IL-12Rß1-deficient patients and two of the 164 kindreds reported. These two CNVs are large deletions found in the heterozygous or homozygous state. We searched for novel families with IL-12Rß1 deficiency due to CNVs. METHODS: We studied six MSMD patients from five unrelated kindreds displaying adverse reactions to BCG vaccination. Three of the patients also presented systemic salmonellosis, two had mucocutaneous candidiasis, and one had disseminated histoplasmosis. We searched for CNVs and other variations by IL12RB1-targeted next-generation sequencing (NGS). RESULTS: We identified six new IL-12Rß1-deficient patients with a complete loss of IL-12Rß1 expression on phytohemagglutinin-activated T cells and/or EBV-transformed B cells. The cells of these patients did not respond to IL-12 and IL-23. Five different CNVs encompassing IL12RB1 (four deletions and one duplication) were identified in these patients by NGS coverage analysis, either in the homozygous state (n = 1) or in trans (n = 4) with a single-nucleotide variation (n = 3) or a small indel (n = 1). Seven of the nine mutations are novel. Interestingly, four of the five CNVs were predicted to be driven by nearby Alu elements, as well as the two previously reported large deletions. The IL12RB1 locus is actually enriched in Alu elements (44.7%), when compared with the rest of the genome (10.5%). CONCLUSION: The IL12RB1 locus is Alu-enriched and therefore prone to rearrangements at various positions. CNVs should be considered in the genetic diagnosis of IL-12Rß1 deficiency.


Subject(s)
Alu Elements , DNA Copy Number Variations , Genetic Association Studies , Genetic Predisposition to Disease , Interleukin-12 Receptor beta 1 Subunit/deficiency , Alleles , Base Sequence , Chromosome Mapping , Female , Gene Expression , Humans , Interferon-gamma , Male , Mutation , Mycobacterium Infections/diagnosis , Mycobacterium Infections/etiology , Mycobacterium Infections/metabolism , Pedigree , Phenotype
19.
Pediatr Blood Cancer ; 64(6)2017 06.
Article in English | MEDLINE | ID: mdl-27873456

ABSTRACT

Mutations of the IL12B and IL12RB1 genes underlie the development of IL-12 p40 and IL-12Rß1 deficiencies, respectively, both of which cause predisposition to infection with weakly virulent mycobacteria and Salmonella. Infections with other intramacrophagic organisms have only been rarely observed. We identified two patients with visceral leishmaniasis who had autosomal recessive IL-12 p40 and IL-12Rß1 deficiencies, respectively. This finding demonstrates the importance of IFN-γ immunity in the control of leishmaniasis. We also searched the literature for similar reports in patients with these and other primary immunodeficiencies.


Subject(s)
Genetic Diseases, Inborn , Immunologic Deficiency Syndromes , Interleukin-12 Subunit p40/deficiency , Leishmaniasis, Visceral , Receptors, Interleukin-12/deficiency , Adolescent , Child , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Male
SELECTION OF CITATIONS
SEARCH DETAIL